CN117815387A - Combination pharmaceutical composition of CDK4/6 inhibitor and anti-PD-L1 antibody - Google Patents

Combination pharmaceutical composition of CDK4/6 inhibitor and anti-PD-L1 antibody Download PDF

Info

Publication number
CN117815387A
CN117815387A CN202311064786.9A CN202311064786A CN117815387A CN 117815387 A CN117815387 A CN 117815387A CN 202311064786 A CN202311064786 A CN 202311064786A CN 117815387 A CN117815387 A CN 117815387A
Authority
CN
China
Prior art keywords
antibody
pharmaceutical composition
formula
compound
present disclosure
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202311064786.9A
Other languages
Chinese (zh)
Inventor
王欢
于鼎
王训强
张喜全
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Original Assignee
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chia Tai Tianqing Pharmaceutical Group Co Ltd filed Critical Chia Tai Tianqing Pharmaceutical Group Co Ltd
Publication of CN117815387A publication Critical patent/CN117815387A/en
Pending legal-status Critical Current

Links

Abstract

The present disclosure relates to a combination pharmaceutical composition of a CDK4/6 inhibitor and an anti-PD-L1 antibody and its use for the treatment of digestive system tumors, in particular gastric or liver cancer. The combination pharmaceutical compositions of the present disclosure produce better efficacy in reducing tumor growth or even eliminating tumors as compared to either drug alone in the combination.

Description

Combination pharmaceutical composition of CDK4/6 inhibitor and anti-PD-L1 antibody
Cross Reference to Related Applications
The present disclosure claims the benefit and priority of chinese invention patent application 202211014692.6 filed at 2022, 23/08 to the national intellectual property agency of the people's republic, the entire contents of which are hereby incorporated by reference herein in their entirety.
Technical Field
The present disclosure is in the field of medicine, and relates to a pharmaceutical combination of a CDK4/6 inhibitor and an anti-PD-L1 antibody, and its use for treating digestive system tumors, in particular gastric or liver cancer.
Background
Cyclin-dependent kinase (CDK) 4/6 is a key regulator of the cell cycle, triggering the cell cycle from the growth phase (G1 phase) to the DNA replication phase (S1 phase). During cell proliferation, the complex formed by CDK4/6 and cyclin D (CyclinD) is capable of phosphorylating retinoblastoma protein (Rb). Once phosphorylated, tumor suppressor Rb releases its tightly bound transcription factor E2F in the unphosphorylated state, E2F activation further transcribes pushing the cell cycle through the restriction site (R-site) and progresses from G1 phase to S phase. Therefore, the inhibition of CDK4/6 makes it impossible to form a CyclinD-CDK4/6 complex, and can block the progress of the cell cycle from the G1 phase to the S phase, thereby achieving the purpose of inhibiting tumor proliferation. WO2016141881 discloses substituted 2-hydro-pyrazole derivatives as selective CDK4/6 inhibitors and in particular discloses compounds of formula (I) of the following structure.
PD-L1 (Programmed deth-ligand), also known as CD247 and B7-H1, is a ligand for Programmed death molecule L (PD-1). PD-L1 is highly expressed on the surfaces of various tumor cells, and the malignancy of tumors and poor prognosis are closely related to the expression level of PD-L1. In the tumor microenvironment, the PD-L1 on the surface of the cancer cells inhibits the activation and proliferation of T cells through the combination with the PD-1 or CD80 on the surface of the T cells, promotes the effector T cells to enter a failure or non-reaction state, induces the apoptosis of the T cells, stimulates the helper T cells to differentiate into regulatory T cells, and further prevents the T cells from killing the tumor cells. The anti-PD-L1 antibody can prevent the related negative regulation signals from being started and conducted by blocking the interaction of PD-L1, PD-1 and CD80, so that the activity of effector T cells in a tumor microenvironment is prevented from being inhibited, and the T cells can play a role in killing and inhibiting tumor cells. The anti-PD-L1 antibody can directly act on tumor tissues, so that the anti-PD-L1 antibody has higher specificity and safety.
Despite the many therapeutic options available for patients with proliferative diseases (e.g., cancer), there remains a need for more effective therapeutic agents for clinical use, particularly in combination with more than one drug.
Disclosure of Invention
In one aspect, the present disclosure provides a pharmaceutical combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and an anti-PD-L1 antibody
In another aspect, the present disclosure provides a pharmaceutical combination for preventing or treating a tumor of the digestive system, comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and an anti-PD-L1 antibody.
In some embodiments of the disclosure, the anti-PD-L1 antibody comprises the following amino acid sequence: a heavy chain CDR1 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 1 or SEQ ID NO. 4; a heavy chain CDR2 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 2 or SEQ ID NO. 5; a heavy chain CDR3 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 3 or SEQ ID NO. 6; a light chain CDR1 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 7 or SEQ ID NO. 10; a light chain CDR2 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 8 or SEQ ID NO. 11; a light chain CDR3 region having at least 80% homology with the amino acid sequence shown in SEQ ID NO. 9 or SEQ ID NO. 12. Further, the anti-PD-L1 antibody comprises the following amino acid sequence: a heavy chain CDR1 region selected from SEQ ID NO. 1 or SEQ ID NO. 4; a heavy chain CDR2 region selected from SEQ ID NO. 2 or SEQ ID NO. 5; a heavy chain CDR3 region selected from SEQ ID NO. 3 or SEQ ID NO. 6; a light chain CDR1 region selected from SEQ ID NO. 7 or SEQ ID NO. 10; a light chain CDR2 region selected from SEQ ID NO. 8 or SEQ ID NO. 11; a light chain CDR3 region selected from SEQ ID NO 9 or SEQ ID NO 12. Still further, the anti-PD-L1 antibody comprises: a heavy chain CDR1 region having the amino acid sequence shown in SEQ ID NO. 1, a heavy chain CDR2 region having the amino acid sequence shown in SEQ ID NO. 2, a heavy chain CDR3 region having the amino acid sequence shown in SEQ ID NO. 3; and a light chain CDR1 region having the amino acid sequence shown in SEQ ID NO. 7, a light chain CDR2 region having the amino acid sequence shown in SEQ ID NO. 8, and a light chain CDR3 region having the amino acid sequence shown in SEQ ID NO. 9. Further, the anti-PD-L1 antibody comprises the following amino acid sequence: a heavy chain variable region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 13 or SEQ ID NO. 14; a light chain variable region having at least 80% homology with the amino acid sequence shown in SEQ ID NO. 15 or SEQ ID NO. 16. Still further, the anti-PD-L1 antibody comprises: a variable heavy chain selected from the group consisting of hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 humanized antibodies, and a variable light chain selected from the group consisting of hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 humanized antibodies.
In some embodiments of the present disclosure, the pharmaceutical combination wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof and the anti-PD-L1 antibody are packaged in the same kit, the kit further comprising instructions for the combined use of the compound of formula (I) or a pharmaceutically acceptable salt thereof and the anti-PD-L1 antibody to treat or prevent a tumor in the digestive system.
In some embodiments, the pharmaceutical combination, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is packaged separately from the anti-PD-L1 antibody in a separate kit, the kit further comprising instructions for use of the compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with the anti-PD-L1 antibody in treating a tumor in the digestive system of a patient.
In some embodiments of the present disclosure, the pharmaceutical combination comprises a pharmaceutical composition of a compound of formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutical composition of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination, wherein the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and the pharmaceutical composition of the anti-PD-L1 antibody are packaged in the same kit, the kit further comprising instructions for the combined use of the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and the pharmaceutical composition of the anti-PD-L1 antibody to treat or prevent a tumor in the digestive tract.
In some embodiments of the present disclosure, the pharmaceutical combination, wherein the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and the pharmaceutical composition of the anti-PD-L1 antibody are packaged separately in separate kits, the kit further comprising instructions for the combined use of the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and the pharmaceutical composition of the anti-PD-L1 antibody to treat or prevent a digestive tract tumor.
In some aspects of the disclosure, the pharmaceutical combination wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof and the anti-PD-L1 antibody are each in the form of a pharmaceutical composition may be administered simultaneously, sequentially or at intervals.
In some embodiments of the present disclosure, the pharmaceutically acceptable salt of the compound of formula (I) is a maleate salt, e.g., a mono-maleate salt of the compound of formula (I).
In some embodiments of the present disclosure, the pharmaceutical combination contains 20 to 240mg, 40 to 180mg, 60 to 180mg, 80 to 180mg, 100 to 180mg, 120 to 180mg, or 150 to 180mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as the compound of formula (I).
In some embodiments of the present disclosure, the pharmaceutical combination contains 20mg, 40mg, 60mg, 80mg, 100mg, 120mg, 150mg, 180mg and/or 240mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as the compound of formula (I).
In some embodiments of the present disclosure, the pharmaceutical combination contains 120mg, 150mg and/or 180mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as the compound of formula (I).
In some embodiments of the present disclosure, the pharmaceutical combination contains 180mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, as the compound of formula (I).
In some embodiments of the present disclosure, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a single dose or multiple doses.
In some embodiments of the present disclosure, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a multi-dose.
In some embodiments of the present disclosure, the pharmaceutical combination, the compound of formula (I) or a pharmaceutically acceptable salt thereof is present in an amount of one daily dose.
In some embodiments of the present disclosure, the pharmaceutical combination, the compound of formula (I) or a pharmaceutically acceptable salt thereof is present in an amount of once daily dose.
In some embodiments of the disclosure, the pharmaceutical combination comprises about 20mg to about 2400mg of the anti-PD-L1 antibody.
In some embodiments of the disclosure, the pharmaceutical combination comprises about 600mg to about 2400mg of the anti-PD-L1 antibody.
In some embodiments of the disclosure, the pharmaceutical combination comprises about 1000mg to about 1500mg of the anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination comprises about 100mg, about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, or about 2400mg of the anti-PD-L1 antibody.
In some embodiments of the disclosure, the pharmaceutical combination comprises about 1200mg of the anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination contains 600 to 2400mg of the pharmaceutical composition of the anti-PD-L1 antibody.
In some embodiments of the disclosure, the pharmaceutical combination contains 600mg, 900mg, 1200mg, 1500mg, 1800mg, 2100mg, 2400mg, or a pharmaceutical composition of an anti-PD-L1 antibody in a range formed by any of the above values.
In some embodiments of the present disclosure, the pharmaceutical combination contains 600 to 2100mg, or 900 to 1500mg, of the pharmaceutical composition of the anti-PD-L1 antibody.
In some embodiments of the disclosure, the pharmaceutical composition of the anti-PD-L1 antibody is a single dose or multiple doses.
In some embodiments of the disclosure, the pharmaceutical composition of the anti-PD-L1 antibody is a multi-dose pharmaceutical composition.
In some embodiments of the disclosure, the pharmaceutical combination, the anti-PD-L1 antibody is present in an amount of one daily dose.
In some embodiments of the present disclosure, the pharmaceutical combination comprises 20 to 240mg, 60 to 180mg, 80 to 180mg, 100 to 180mg, 120 to 180mg, or 150 to 180mg of a pharmaceutical composition of a compound of formula (I) or a pharmaceutically acceptable salt thereof, and 600 to 2400mg of a pharmaceutical composition of an anti-PD-L1 antibody, wherein the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is single dose or multi-dose, wherein the pharmaceutical composition of the anti-PD-L1 antibody is single dose or multi-dose, calculated as the compound of formula (I); preferably, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a multi-dose, and the pharmaceutical composition of the anti-PD-L1 antibody is a multi-dose.
In some embodiments of the present disclosure, the pharmaceutical combination comprises 20mg, 40mg, 60mg, 80mg, 100mg, 120mg, 150mg and/or 180mg of a pharmaceutical composition of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and 600mg, 900mg, 1200mg, 1500mg, 1800mg, 2100mg, 2400mg, or a range of any of the above values of an anti-PD-L1 antibody, wherein the pharmaceutical composition of the compound of formula (I), or a pharmaceutically acceptable salt thereof, is single dose or multiple doses, wherein the pharmaceutical composition of the anti-PD-L1 antibody is single dose or multiple doses; preferably, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a multi-dose, and the pharmaceutical composition of the anti-PD-L1 antibody is a multi-dose.
In some embodiments of the present disclosure, the pharmaceutical combination comprises 60mg, 80mg, 100mg, 120mg, 150mg and/or 180mg of the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and 600 to 2100mg, or 900 to 1500mg of the pharmaceutical composition of the anti-PD-L1 antibody, calculated as the compound of formula (I), wherein the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a single dose or multiple doses, wherein the pharmaceutical composition of the anti-PD-L1 antibody is a single dose or multiple doses; preferably, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a multi-dose, and the pharmaceutical composition of the anti-PD-L1 antibody is a multi-dose.
In some embodiments of the present disclosure, the pharmaceutical combination comprising 120mg, 150mg and/or 180mg of the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and 1200mg of the pharmaceutical composition of the anti-PD-L1 antibody, calculated as the compound of formula (I), wherein the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a single dose or multiple doses, wherein the pharmaceutical composition of the anti-PD-L1 antibody is a single dose or multiple doses; preferably, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a multi-dose, and the pharmaceutical composition of the anti-PD-L1 antibody is a multi-dose.
In some embodiments of the present disclosure, the pharmaceutical combination comprising 180mg of the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof and 1200mg of the pharmaceutical composition of the anti-PD-L1 antibody, calculated as the compound of formula (I), wherein the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a single dose or multiple doses, wherein the pharmaceutical composition of the anti-PD-L1 antibody is a single dose or multiple doses; preferably, the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof is a multi-dose, and the pharmaceutical composition of the anti-PD-L1 antibody is a multi-dose.
In some embodiments of the present disclosure, the pharmaceutical combination, which is a pharmaceutical composition suitable for administration over a single treatment cycle (e.g., one treatment cycle of 21 days or 28 days), comprises a pharmaceutical composition comprising 1260 to 5040mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, calculated as a compound of formula (I) and a pharmaceutical composition comprising 600 to 2400mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination, which is a pharmaceutical composition suitable for administration over a single treatment cycle (e.g., one treatment cycle of 21 days or 28 days), comprises a pharmaceutical composition comprising 2520 to 3780mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, based on the compound of formula (I) and a pharmaceutical composition comprising 900 to 1500mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination, which is a pharmaceutical composition suitable for administration over a single treatment cycle (e.g., 21 days of a treatment cycle), comprises a pharmaceutical composition comprising 2520mg, 3150mg, or 3780mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as the compound of formula (I) and a pharmaceutical composition comprising 1200mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination, which is a pharmaceutical composition suitable for administration over a single treatment cycle (e.g., 21 days of one treatment cycle), comprises a pharmaceutical composition comprising 3780mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, as the compound of formula (I) and a pharmaceutical composition comprising 1200mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the pharmaceutical combination is a pharmaceutical composition suitable for administration over a single treatment period (e.g., one treatment period of 21 days or 28 days) comprising (1.0-6.5) by weight: 1. preferably (2.0-6.5): 1, more preferably (2.0-3.5): 1, more preferably (2.1-3.2): 1, most preferably (2.6-3.2): 1, a compound of formula (I) or a pharmaceutically acceptable salt thereof, and an anti-PD-L1 antibody. Wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof and the anti-PD-L1 antibody are packaged separately or together.
In some embodiments of the present disclosure, the kits of the present disclosure are kits suitable for administration over a single treatment cycle (e.g., one treatment cycle of 21 days or 28 days), including pharmaceutical compositions comprising 1260 to 5040mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as calculated as a compound of formula (I) and pharmaceutical compositions comprising 600 to 2400mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the kits of the present disclosure are kits suitable for administration over a single treatment cycle (e.g., one treatment cycle of 21 days or 28 days), including pharmaceutical compositions comprising 2520-3780 mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as a compound of formula (I) and pharmaceutical compositions comprising 900-1500 mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the kits of the present disclosure are kits suitable for administration over a single treatment cycle (e.g., 21 days of a treatment cycle), including a pharmaceutical composition comprising 2520mg, 3150mg, or 3780mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as the compound of formula (I) and a pharmaceutical composition comprising 1200mg of an anti-PD-L1 antibody.
In some embodiments of the present disclosure, the kits of the present disclosure are kits suitable for administration over a single treatment cycle (e.g., 21 days of a treatment cycle), including a pharmaceutical composition comprising 3780mg of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as a compound of formula (I) and a pharmaceutical composition comprising 1200mg of an anti-PD-L1 antibody.
In another aspect, the present disclosure provides a kit comprising a pharmaceutical combination as described herein, further comprising instructions for the combined use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and an anti-PD-L1 antibody to treat or prevent a tumor in the digestive system.
In another aspect, the present disclosure provides a kit comprising a pharmaceutical combination as described herein, further comprising instructions for the combined use of a pharmaceutical composition of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutical composition of an anti-PD-L1 antibody to treat or prevent a tumor in the digestive system.
Use, administration/treatment method of pharmaceutical combination
In another aspect, the present disclosure also provides a method of treating or preventing a tumor of the digestive system, comprising administering to an individual in need thereof a therapeutically effective amount of a pharmaceutical combination as described in the present disclosure.
In another aspect, the present disclosure also provides the use of a pharmaceutical combination according to the present disclosure for the manufacture of a medicament for the treatment or prevention of a tumor of the digestive system.
In another aspect, the present disclosure also provides the use of a pharmaceutical combination as described in the present disclosure for the treatment or prevention of a tumor of the digestive system.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the anti-PD-L1 antibody are each in the form of a pharmaceutical composition for simultaneous, sequential or separate administration.
In some embodiments of the present disclosure, in the above use or method, the compound of formula (I) or a pharmaceutically acceptable salt thereof and the anti-PD-L1 antibody are each administered in a separate administration. In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the anti-PD-L1 antibody are administered separately on the same or different dosing schedules. In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the anti-PD-L1 antibody are administered separately on different dosing schedules.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered at a dose of 60mg, 120mg, 150mg, or 180mg once daily.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered at a dose of 120mg, 150mg, or 180mg once daily.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered at a dose of 180mg once daily.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered as a continuous daily administration.
In some embodiments of the disclosure, the anti-PD-L1 antibody is administered at a dose of 600 to 2100mg, or 900 to 1500mg, once daily.
In some embodiments of the disclosure, the anti-PD-L1 antibody is administered at a dose of 1200mg once daily in the use or method.
In some embodiments of the disclosure, the anti-PD-L1 antibody is administered on the first day of each cycle.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the anti-PD-L1 antibody have the same or different treatment cycles.
In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the anti-PD-L1 antibody have the same treatment cycle, e.g., one treatment cycle every 1 week, every 2 weeks, every 3 weeks, or every 4 weeks. In some embodiments of the present disclosure, the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the anti-PD-L1 antibody have different treatment cycles in the use or method.
In some embodiments of the present disclosure, the use or method wherein 21 days is one treatment cycle, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered daily on days 1-21 of each cycle, and the anti-PD-L1 antibody is administered on the first day of each cycle. In a specific embodiment, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered once daily on days 1-21 of each cycle, and the anti-PD-L1 antibody is administered once on the first day of each cycle.
In some embodiments of the disclosure, the use or method, wherein the anti-PD-L1 antibody can comprise administering to the subject a dose selected from 0.01 to 40mg/kg,0.1 to 30mg/kg,0.1 to 20mg/kg,0.1 to 15mg/kg,0.1 to 10mg/kg,1 to 15mg/kg,1 to 20mg/kg,1 to 3mg/kg,3 to 10mg/kg,3 to 15mg/kg,3 to 20mg/kg,3 to 30mg/kg,10 to 20mg/kg, or 15 to 20 mg/kg; or from 60mg to 2400mg,90mg to about 1500mg, 120mg to 1500mg,300mg to 900mg,600mg to 900mg,300mg to 1200mg, or 900mg to 1200 mg.
In some embodiments of the present disclosure, the use or method, 21 days is a treatment cycle, 20 to 240mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered daily on days 1-21 of each cycle, and 600 to 2400mg of the anti-PD-L1 antibody is administered on the first day of each cycle.
In some embodiments of the present disclosure, the use or method, 21 days is a treatment cycle, 60 to 180mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered daily on days 1-21 of each cycle, and 900 to 1500mg of the anti-PD-L1 antibody is administered on the first day of each cycle.
In some embodiments of the present disclosure, the use or method, 21 days is a treatment cycle, 120mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered daily on days 1-21 of each cycle, and 1200mg of the anti-PD-L1 antibody is administered on the first day of each cycle.
In some embodiments of the present disclosure, the use or method, wherein 21 days is one treatment cycle, 150mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered daily on days 1-21 of each cycle, and 1200mg of the anti-PD-L1 antibody is administered on the first day of each cycle.
In some embodiments of the present disclosure, the use or method, wherein 21 days is one treatment cycle, 180mg of the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered daily on days 1-21 of each cycle, and 1200mg of the anti-PD-L1 antibody is administered on the first day of each cycle.
In some embodiments of the present disclosure, at one treatment cycle every 21 days, at (1.0-6.5), respectively: 1. preferably (2.0-6.5): 1, more preferably (2.0-3.5): 1, more preferably (2.1-3.2): 1, most preferably (2.6-3.2): 1, by weight ratio to the subject, a compound of formula (I) or a pharmaceutically acceptable salt thereof and an anti-PD-L1 antibody, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof and the anti-PD-L1 antibody are administered in multiple doses and multiple doses, respectively.
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure is an advanced and/or metastatic digestive system tumor.
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure is an advanced and/or metastatic digestive system tumor that failed standard treatment.
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure has an abnormality in the cell cycle signal pathway comprising: high expression of Cyclin D1, CDK4/6 mutation, etc.
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure is an imagewise confirmed unresectable digestive system tumor.
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure is a non-resectable advanced and/or metastatic digestive system tumor that fails standard treatment.
In other embodiments of the present disclosure, the digestive system tumor of the present disclosure is a digestive system tumor resistant to an immune checkpoint inhibitor.
In other embodiments of the present disclosure, the immune checkpoint inhibitor of the present disclosure is PD-1/PD-L1 mab.
In some embodiments of the disclosure, the digestive system tumor of the disclosure is a PD-1/PD-L1 mab-resistant digestive system tumor.
In some embodiments of the disclosure, the digestive system tumor of the disclosure is a PD-1/PD-L1 mab-resistant digestive system tumor in which there is an abnormality in the cell cycle signal pathway.
In some embodiments of the disclosure, the digestive system tumor of the disclosure is a PD-1/PD-L1 mab-resistant, unresectable digestive system tumor that has an abnormal cell cycle signal pathway.
In some embodiments of the disclosure, the digestive system tumor of the disclosure is a PD-1/PD-L1 mab-resistant, unresectable advanced and/or metastatic digestive system tumor that has abnormal cell cycle signaling pathways and failed standard treatment.
In other embodiments of the present disclosure, the digestive system tumor of the present disclosure is selected from a digestive system tumor that has previously received one or more treatments (e.g., failed treatment) of paclitaxel, gemcitabine, cisplatin, nedaplatin, carlizumab, an Luoti ni, afatinib, tirelib, terlipp Li Shan antibody, or radiation therapy.
In other embodiments of the present disclosure, the digestive system tumor of the present disclosure is selected from a digestive system tumor that has previously received one or more treatments (e.g., treatment failure) of paclitaxel + cisplatin, gemcitabine + cisplatin, carlizumab + An Luoti ni, albumin paclitaxel + nedaplatin + tirelib, paclitaxel + cisplatin + radiation therapy, afatinib + terlipp Li Shan antibody, or albumin paclitaxel + nedaplatin + tirelib.
In other embodiments of the present disclosure, the digestive system tumor of the present disclosure is selected from a digestive system tumor that has previously received treatment (e.g., failed treatment) with paclitaxel + cisplatin, gemcitabine + cisplatin, and carlizumab + An Luoti, or that has previously received treatment (e.g., failed treatment) with albumin paclitaxel + nedaplatin + tirelib, paclitaxel + cisplatin + radiation therapy, afatinib + terlipp Li Shan antibody, and albumin paclitaxel + nedaplatin + tirelib.
In other embodiments of the present disclosure, the digestive system tumor of the present disclosure is selected from a digestive system tumor that has been previously treated with paclitaxel + cisplatin, gemcitabine + cisplatin, and carlizumab + An Luoti in sequence (e.g., failed treatment), or that has been previously treated with albumin paclitaxel + nedaplatin + tirelib, paclitaxel + cisplatin + radiation therapy, afatinib + terlipp Li Shan antibody, and albumin paclitaxel + nedaplatin + tirelib in sequence (e.g., failed treatment).
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure is selected from esophageal cancer, gastric cancer, colorectal cancer, or liver cancer.
In some embodiments of the present disclosure, the digestive system tumor of the present disclosure is selected from esophageal cancer.
In other embodiments of the present disclosure, the esophageal cancer of the present disclosure is esophageal cancer that failed standard treatment and/or failed immune checkpoint inhibitor treatment.
In other embodiments of the present disclosure, the esophageal cancer of the present disclosure is esophageal cancer that failed standard treatment and/or is resistant to PD-1/PD-L1 mab.
In other embodiments of the present disclosure, the esophageal cancer of the present disclosure is selected from esophageal squamous carcinoma.
In other embodiments of the present disclosure, the esophageal squamous carcinoma of the present disclosure is selected from esophageal squamous carcinoma that had previously been treated with paclitaxel + cisplatin, gemcitabine + cisplatin, and carlizumab + An Luoti in sequence (e.g., failed treatment), or that had previously been treated with albumin paclitaxel + nedaplatin + tirelib, paclitaxel + cisplatin + radiation therapy, afatinib + terlipp Li Shan antibody, and albumin paclitaxel + nedaplatin + tirelib in sequence (e.g., failed treatment).
In some embodiments of the present disclosure, the standard treatment of the present disclosure includes surgical treatment, radiation treatment, targeted treatment, chemotherapy, and immunotherapy.
In other embodiments of the present disclosure, the standard therapy of the present disclosure is selected from paclitaxel therapy, gemcitabine therapy, cisplatin therapy, nedaplatin therapy, an Luoti ni therapy, afatinib therapy, or radiation therapy.
In other embodiments of the present disclosure, the PD-1/PD-L1 mab of the present disclosure is selected from the group consisting of carlizumab, tirelimumab, and terlipressin Li Shan.
CDK4/6 inhibitors
As used in the present disclosure, the CDK4/6 inhibitor may be a compound of formula (I) or a pharmaceutically acceptable salt thereof,
as used herein, a pharmaceutically acceptable salt of a compound of formula (I) may be a maleate salt of a compound of formula (I) (e.g., a mono-maleate salt of a compound of formula (I)). The dosages of the compounds of formula (I) or pharmaceutically acceptable salts thereof referred to in this disclosure are based on the molecular weight of the compounds of formula (I) unless otherwise indicated.
The compounds of formula (I) or pharmaceutically acceptable salts thereof used in the present disclosure may be prepared by methods of the prior art, for example by methods described in reference to WO 2016141881.
Pharmaceutical compositions of compounds of formula (I) or pharmaceutically acceptable salts thereof
In some embodiments of the present disclosure, a single dose of the pharmaceutical composition of the compound of formula (I) or a pharmaceutically acceptable salt thereof comprises 50mg or 60mg.
In some embodiments of the present disclosure, the total dose of the compound of formula (I), or a pharmaceutically acceptable salt thereof, administered daily on days 1-21 of each cycle comprises 1260-5040 mg, per 21-day treatment cycle. In some embodiments, the total dose of the compound of formula (I) or a pharmaceutically acceptable salt thereof comprises a range selected from 1260mg, 2520mg, 3150mg, 3780mg, 5040mg, or any of the above. In some embodiments, the total dose of the compound of formula (I) or a pharmaceutically acceptable salt thereof preferably comprises 2520mg to 3780mg.
In some embodiments, the pharmaceutical compositions include, but are not limited to, formulations suitable for oral, parenteral, topical administration; in some embodiments, the pharmaceutical composition is a formulation suitable for oral administration; in some embodiments, the pharmaceutical composition is a solid formulation suitable for oral administration; in some embodiments, the pharmaceutical composition includes, but is not limited to, a tablet, a capsule.
The pharmaceutical compositions of the compounds of formula (I) or pharmaceutically acceptable salts thereof of the present disclosure may be manufactured using methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, freeze-drying, and the like.
The solid oral compositions may be prepared by conventional mixing, filling or tabletting methods. For example, it can be obtained by the following method: the active compound is mixed with solid auxiliary materials, the resulting mixture is optionally milled, if desired with other suitable auxiliary materials, and the mixture is then processed to granules, giving a tablet or dragee core. Suitable excipients include, but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
anti-PD-L1 antibodies
The anti-PD-L1 antibodies described in the present disclosure comprise: HCDR1 of the amino acid sequence shown in SEQ ID NO. 1 or 7, HCDR2 of the amino acid sequence shown in SEQ ID NO. 2 or 8, HCDR3 of the amino acid sequence shown in SEQ ID NO. 3 or 9, LCDR1 of the amino acid sequence shown in SEQ ID NO. 4 or 10, LCDR2 of the amino acid sequence shown in SEQ ID NO. 5 or 11, and LCDR3 of the amino acid sequence shown in SEQ ID NO. 6 or 12.
In some embodiments, the anti-PD-L1 antibody comprises: HCDR1 of the amino acid sequence shown in SEQ ID NO. 1, HCDR2 of the amino acid sequence shown in SEQ ID NO. 2, HCDR3 of the amino acid sequence shown in SEQ ID NO. 3, LCDR1 of the amino acid sequence shown in SEQ ID NO. 4, LCDR2 of the amino acid sequence shown in SEQ ID NO. 5, and LCDR3 of the amino acid sequence shown in SEQ ID NO. 6; or HCDR1 of the amino acid sequence shown in SEQ ID NO. 7, HCDR2 of the amino acid sequence shown in SEQ ID NO. 8, HCDR3 of the amino acid sequence shown in SEQ ID NO. 9, LCDR1 of the amino acid sequence shown in SEQ ID NO. 10, LCDR2 of the amino acid sequence shown in SEQ ID NO. 11, and LCDR3 of the amino acid sequence shown in SEQ ID NO. 12. The anti-PD-L1 antibody CDR sequences are provided in Table 1 below.
Table 1.anti-PD-L1 antibodiesCDR sequences
It will be understood by those of skill in the art that unless otherwise specified, the terms "CDR" and "complementarity determining region" of a given antibody or region thereof (e.g., variable region) are understood to encompass complementarity determining regions defined by any known scheme. Although CDR regions have been shown in table 1, where reference is made to defining antibodies with specific CDR sequences defined by certain divisions of the present disclosure, the scope of the antibodies also encompasses antibodies that are converted to CDR sequence definitions of other arbitrary definitions (e.g., combinations of one or more of the Kabat, chothia, IMGT or Contact etc. definitions as known in the art).
In some embodiments, the anti-PD-L1 antibody comprises a heavy chain variable region having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 13 or 15, and a light chain variable region having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 14 or 16. In some embodiments, the anti-PD-L1 antibody comprises a heavy chain variable region of the amino acid sequence set forth in SEQ ID NO. 13 or 15 and a light chain variable region of the amino acid sequence set forth in SEQ ID NO. 14 or 16. In some embodiments, the anti-PD-L1 antibody comprises a heavy chain variable region having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 13, and a light chain variable region having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 14. In other embodiments, the anti-PD-L1 antibody comprises a heavy chain variable region having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 15, and a light chain variable region having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 16. In some embodiments, the anti-PD-L1 antibody comprises a heavy chain variable region set forth in SEQ ID NO. 13 and a light chain variable region set forth in SEQ ID NO. 14. In other embodiments, the anti-PD-L1 antibody comprises a heavy chain variable region set forth in SEQ ID NO. 15 and a light chain variable region set forth in SEQ ID NO. 16.
QITLKESGPTLVKPTQTLTLTCTVSGFSLSTYGVHWIRQPPGKALEWLGVIWRGVTTDYNAAFMSRL TITKDNSKNQVVLTMNNMDPVDTATYYCARLGFYAMDYWGQGTLVTVSS(SEQ ID NO:13);
DIQMTQSPSSLSASVGDRVTITCKASQSVSNDVAWYQQKPGKAPKLLIYYAANRYTGVPDRFSGSGY GTDFTFTISSLQPEDIATYFCQQDYTSPYTFGQGTKLEIK(SEQ ID NO:14);
EVQLVESGGGLVKPGGSLRLSCAASGFIFRSYGMSWVRQAPGKGLEWVASISSGGSTYYPDSVKGRF TISRDNAKNSLYLQMNSLRAEDTAVYDCARGYDSGFAYWGQGTLVTVSS(SEQ ID NO:15);
DIVLTQSPASLAVSPGQRATITCRASQSVSTSSSSFMHWYQQKPGQPPKLLIKYASNLESGVPARFSGS GSGTDFTLTINPVEANDTANYYCQHSWEIPYTFGQGTKLEIK(SEQ ID NO:16)。
In some embodiments, the anti-PD-L1 antibody may further comprise a constant region of an immunoglobulin, or a fragment, analog, variant, or derivative of the constant region. In some embodiments, the constant region is from a human immunoglobulin heavy chain, such as the heavy chain of IgG1, igG2, igG3, and IgG4 or other classes of immunoglobulins, preferably the heavy chain of IgG 1. In some embodiments, the constant region may comprise any of the modifications described herein, such as amino acid insertions, deletions, substitutions, or chemical modifications. In some embodiments, the constant region comprises a mutation that alters effector function. In some embodiments, any amino acid residue of the constant region may be substituted with an amino acid residue of any allotype (allotype), preferably with an amino acid residue of G1m (3) and/or nG1m (1).
In some embodiments, the anti-PD-L1 antibody comprises a heavy chain having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID No. 17 or 19, and a light chain having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID No. 18 or 20. In some embodiments, the anti-PD-L1 antibody comprises a heavy chain having an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 17 and a light chain having an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 18. In other embodiments, the anti-PD-L1 antibody comprises a heavy chain having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 19, and a light chain having an amino acid sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence set forth in SEQ ID NO. 20. In some embodiments, the anti-PD-L1 antibody comprises a heavy chain as set forth in SEQ ID NO. 17 and a light chain as set forth in SEQ ID NO. 18. In other embodiments, the anti-PD-L1 antibody comprises a heavy chain as set forth in SEQ ID NO. 19 and a light chain as set forth in SEQ ID NO. 20.
QITLKESGPTLVKPTQTLTLTCTVSGFSLSTYGVHWIRQPPGKALEWLGVIWRGVTTDYNAAFMSRLTITKDNSKNQVVLTMNNMDPVDTATYYCARLGFYAMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVAVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK(SEQ ID NO:17);
DIQMTQSPSSLSASVGDRVTITCKASQSVSNDVAWYQQKPGKAPKLLIYYAANRYTGVPDRFSGSGYGTDFTFTISSLQPEDIATYFCQQDYTSPYTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC(SEQ ID NO:18);
EVQLVESGGGLVKPGGSLRLSCAASGFIFRSYGMSWVRQAPGKGLEWVASISSGGSTYYPDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYDCARGYDSGFAYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT KVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVAVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK(SEQ ID NO:19);
DIVLTQSPASLAVSPGQRATITCRASQSVSTSSSSFMHWYQQKPGQPPKLLIKYASNLESGVPARFSGSGSGTDFTLTINPVEANDTANYYCQHSWEIPYTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC(SEQ ID NO:20)。
anti-PD-L1 antibodies or antigen fragments thereof of the present disclosure include, but are not limited to, 13C5, 5G11, ch13C5-hIgG1, ch13C5-hIgG4, ch5G11-hIgG1, ch5G11-hIgG4, hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 monoclonal antibodies described in the published application No. WO2016022630 or CN 107001463.
In one embodiment, the anti-PD-L1 antibody is hu5G11-hIgG1 comprising the heavy chain variable region of SEQ ID NO. 13 and the light chain variable region of SEQ ID NO. 14.
In one embodiment, the anti-PD-L1 antibody is hu5G11-hIgG1 comprising the heavy chain of SEQ ID NO. 17 and the light chain of SEQ ID NO. 18.
Pharmaceutical composition of anti-PD-L1 antibody
In some embodiments of the disclosure, a single dose of the pharmaceutical composition of the anti-PD-L1 antibody comprises 100mg or 600mg of the anti-PD-L1 antibody.
In some embodiments of the present disclosure, the total dose of the pharmaceutical composition of the anti-PD-L1 antibody is 600-2400 mg. In some embodiments, the total dose of the pharmaceutical composition of the anti-PD-L1 antibody comprises a range selected from 600mg, 900mg, 1200mg, 1500mg, 1800mg, 2100mg, 2400mg, or any of the above. In some embodiments, the total dose of the pharmaceutical composition of the anti-PD-L1 antibody is preferably 600-2100 mg, or 900-1500 mg.
In some embodiments of the present disclosure, the pharmaceutical composition of the anti-PD-L1 antibody comprises one or more of a buffer, an isotonicity adjusting agent, a stabilizer, and/or a surfactant. In particular, the pharmaceutical composition of the anti-PD-L1 antibody comprises 1-150mg/mL of the anti-PD-L1 antibody (e.g., monoclonal antibody), 3-50mM buffer, 2-150mg/mL of an isotonicity modifier/stabilizer, and 0.01-0.8mg/mL of a surfactant, and has a pH of about 4.5-6.8.
In some embodiments of the present disclosure, the pharmaceutical composition of the anti-PD-L1 antibody has an anti-PD-L1 mab concentration of about 5-150mg/mL, calculated as w/v; preferably about 10-60mg/mL; more preferably about 10-30mg/mL. In some embodiments, the anti-PD-L1 mab is present in a mass volume concentration of about 10mg/mL, about 20mg/mL, about 30mg/mL, about 40mg/mL, about 50mg/mL, about 60mg/mL, about 70mg/mL, about 80mg/mL, about 90mg/mL, about 100mg/mL, about 110mg/mL, or about 120mg/mL, preferably about 10mg/mL, about 20mg/mL, about 30mg/mL, about 40mg/mL, about 50mg/mL, or about 60mg/mL, more preferably about 10mg/mL, about 20mg/mL, or about 30mg/mL. In some embodiments, the anti-PD-L1 mab mass volume concentration is about 10mg/mL. In other embodiments, the anti-PD-L1 mab is about 30mg/mL in mass volume concentration. In other embodiments, the anti-PD-L1 mab is about 60mg/mL in mass volume concentration.
In some embodiments of the disclosure, the buffer is a histidine salt buffer. The histidine salt buffer concentration is about 5-30mM, preferably about 10-25mM, more preferably about 10-20mM, and most preferably about 10-15mM. In some embodiments, the histidine salt buffer concentration is about 5mM, about 10mM, about 15mM, about 20mM, about 25mM or about 30mM. In some embodiments, the histidine salt buffer concentration is about 10mM. In other embodiments, the histidine salt buffer concentration is about 15mM. In other embodiments, the histidine salt buffer concentration is about 20mM. Wherein the histidine salt buffer comprises histidine and hydrochloric acid.
In some embodiments of the present disclosure, the isotonicity modifier/stabilizer is from about 20 to 150mg/mL sucrose, preferably from about 40 to 100mg/mL sucrose, more preferably from about 60 to 80mg/mL sucrose, calculated as w/v. In some embodiments, the sucrose is at a concentration of about 40mg/mL, 50mg/mL, 60mg/mL, 70mg/mL, 80mg/mL, 90mg/mL, or 100mg/mL. In some embodiments, the sucrose is at a concentration of about 60mg/mL. In some embodiments, the sucrose is at a concentration of about 70mg/mL. In some embodiments, the sucrose is at a concentration of about 80mg/mL. In some embodiments, the sucrose is at a concentration of about 90mg/mL.
In some embodiments of the present disclosure, the surfactant is selected from polysorbate 80, polysorbate 20, poloxamer 188; preferably polysorbate 80 or polysorbate 20; more preferably polysorbate 80. In some embodiments, the surfactant is present at a concentration of about 0.05 to about 0.6mg/mL, preferably about 0.1 to about 0.4mg/mL, and more preferably about 0.2 to about 0.3mg/mL, calculated as w/v.
In some embodiments of the present disclosure, the surfactant is about 0.01-0.8mg/mL polysorbate 80 or polysorbate 20 calculated as w/v. In some embodiments, the surfactant is about 0.05 to about 0.6mg/mL polysorbate 80, preferably about 0.1 to about 0.4mg/mL polysorbate 80, more preferably about 0.2 to about 0.3mg/mL polysorbate 80, and most preferably about 0.2mg/mL polysorbate 80. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.1mg/mL, 0.2mg/mL, 0.3mg/mL, 0.4mg/mL, 0.5mg/mL, or 0.6mg/mL; preferably, the polysorbate 80 content of the pharmaceutical composition is about 0.2mg/mL, 0.3mg/mL, 0.4mg/mL or 0.5mg/mL; more preferably, the polysorbate 80 content of the pharmaceutical composition is about 0.2mg/mL, 0.3mg/mL or 0.4mg/mL; optimally, the polysorbate 80 content of the pharmaceutical composition is about 0.2mg/mL. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.1mg/mL. In other embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.2mg/mL. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.3mg/mL. In other embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.4mg/mL. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.5mg/mL.
In some embodiments of the present disclosure, the aqueous solution of the pharmaceutical composition has a pH value selected from 4.0-6.8; preferably 4.5 to 6.5; more preferably 5.5 to 6.0; most preferably 5.5. In some embodiments, the aqueous pharmaceutical composition has a pH of about 4.5, about 4.8, about 5.0, about 5.2, about 5.4, about 5.5, about 5.6, about 5.8, or about 6.0, preferably about 5.0, about 5.2, about 5.4, about 5.5, or about 5.6, more preferably about 5.5. In some embodiments, the aqueous pharmaceutical composition has a pH of about 5.0. In some embodiments, the aqueous pharmaceutical composition has a pH of about 5.2. In some embodiments, the aqueous pharmaceutical composition has a pH of about 5.4. In some embodiments, the aqueous pharmaceutical composition has a pH of about 5.5. In some embodiments, the aqueous pharmaceutical composition has a pH of about 5.6. In some embodiments, the aqueous pharmaceutical composition has a pH of about 5.8. In some embodiments, the aqueous pharmaceutical composition has a pH of about 6.0.
In some embodiments of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 20mg/mL, (b) sucrose at a mass-volume concentration of about 70mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.1mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.0. In a specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) anti-PD-L1 mab at a mass volume concentration of about 20mg/mL, (b) sucrose at a mass volume concentration of about 70mg/mL, (c) polysorbate 80 at a mass volume concentration of about 0.1mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.0.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 10mg/mL, (b) sucrose at a mass-volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.2mg/mL, (d) histidine at a molar concentration of about 10mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 50mg/mL, (b) sucrose at a mass-volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.3mg/mL, (d) histidine at a molar concentration of about 10mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 100mg/mL, (b) sucrose at a mass-volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.5mg/mL, (d) histidine at a molar concentration of about 10mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 30mg/mL, (b) sucrose at a mass-volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.2mg/mL, (d) histidine at a molar concentration of about 10mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 60mg/mL, (b) sucrose at a mass-volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.2mg/mL, (d) histidine at a molar concentration of about 10mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) an anti-PD-L1 antibody at a mass-volume concentration of about 10mg/mL, (b) sucrose at a mass-volume concentration of about 70mg/mL, (c) polysorbate 80 at a mass-volume concentration of about 0.4mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, an appropriate amount of acetic acid, and adjusting the pH of the composition to about 6.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition comprises: (a) anti-PD-L1 mab at a mass volume concentration of about 10mg/mL, (b) sucrose at a mass volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass volume concentration of about 0.2mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, an appropriate amount of hydrochloric acid, and adjusting the pH of the composition to about 5.5.
In another specific embodiment of the present disclosure, the pharmaceutical composition is a water-soluble injection, including but not limited to a water-soluble formulation that has not been lyophilized or a water-soluble formulation that has been reconstituted from a lyophilized powder. In other embodiments, the pharmaceutical composition is a lyophilized formulation. The lyophilized preparation refers to a preparation prepared by subjecting an aqueous solution to a lyophilization process in which a substance is first frozen, then the amount of solvent is reduced by sublimation (primary drying process) and then the amount of solvent is reduced by desorption (secondary drying process) until the amount of solvent is a value that no longer supports biological activity or chemical reaction. The lyophilized formulations of the present disclosure may also be dried by other methods known in the art, such as spray drying and bubble drying (bubbledrying).
Mode of administration
The following is not intended to limit the manner of administration of the pharmaceutical combinations of the present disclosure.
The components of the pharmaceutical compositions of the present disclosure may each be administered independently, or some or all of them together, in a suitable variety of ways, including, but not limited to, orally or parenterally (via intravenous, intramuscular, topical, or subcutaneous routes). In some embodiments, the components of the pharmaceutical combinations of the present disclosure may each be administered orally or by injection, e.g., intravenously or intraperitoneally, independently, or in combination with some or all of them.
The components of the pharmaceutical compositions of the present disclosure may each independently, or some or all of them together, be in suitable dosage forms including, but not limited to, tablets, troches, pills, capsules (e.g., hard, soft, enteric, microcapsules), elixirs, granules, syrups, injections (intramuscular, intravenous, intraperitoneal), granules, emulsions, suspensions, solutions, dispersions, and dosage forms of sustained release formulations for oral or non-oral administration.
The components of the pharmaceutical combinations of the present disclosure may each independently, or some or all of them together contain a pharmaceutically acceptable carrier and/or excipient.
The pharmaceutical combinations of the present disclosure may also comprise additional therapeutic agents. In one embodiment, the additional therapeutic agent may be a proliferative disease therapeutic agent known in the art.
Technical effects
Generally, the use of the pharmaceutical combinations of the present disclosure described above will help:
(1) Producing a better therapeutic effect in reducing the growth of or even eliminating the tumor than either drug administered alone in the combination;
(2) Providing a smaller amount of administration compared to either drug administered alone in the combination;
(3) Providing a treatment with good tolerance in the patient with fewer adverse reactions and/or complications than either drug administered alone;
(4) Providing better disease control rate among treated patients;
(5) Providing a longer survival (e.g., median survival, progression free survival, or total survival) in the treated patient;
(6) Providing a longer survival (e.g., median survival, progression free survival, or total survival) for the treated patient compared to standard chemotherapy;
(7) Provide longer duration of disease remission (DOR); and/or
(8) Compared with any one of the medicines singly administered in the combination, the composition has good activity of treating tumor or proliferative diseases and shows more excellent anti-tumor synergistic effect.
Definition and description
The following terms used in this disclosure have the following meanings, unless otherwise indicated. A particular term, unless otherwise defined, shall not be construed as being ambiguous or otherwise unclear, but shall be construed in accordance with the ordinary meaning in the art. When trade names are presented in this disclosure, it is intended to refer to their corresponding commercial products or active ingredients thereof.
As used herein, the term "pharmaceutical combination" refers to a combination of two or more active ingredients administered simultaneously or sequentially (either as the respective active ingredients themselves or as derivatives, prodrugs or compositions of their respective pharmaceutically acceptable salts or esters). The terms "pharmaceutical combination" and "combination pharmaceutical composition" are used interchangeably herein.
The term "antibody" refers to a binding protein having at least one antigen binding domain. The antibodies and fragments thereof of the present disclosure may be whole antibodies or any fragment thereof. Thus, antibodies and fragments of the present disclosure include monoclonal antibodies or fragments thereof and antibody variants or fragments thereof, as well as immunoconjugates. Examples of antibody fragments include Fab fragments, fab 'fragments, F (ab') 2 fragments, fv fragments, isolated CDR regions, single chain Fv molecules (scFv), fd fragments and other antibody fragments known in the art. Antibodies and fragments thereof may also include recombinant polypeptides, fusion proteins, and bispecific antibodies. The anti-PD-L1 antibodies and fragments thereof disclosed herein may be of the IgG1, igG2, igG3 or IgG4 isotype. The term "isotype" refers to the type of antibody encoded by the heavy chain constant region gene. In one embodiment, the anti-PD-L1 antibodies and fragments thereof disclosed herein are of the IgG1 or IgG4 isotype. The PD-L1 antibodies and fragments thereof of the present disclosure may be derived from any species including, but not limited to, mice, rats, rabbits, primates, llamas, and humans. The PD-L1 antibody and fragments thereof may be chimeric, humanized or fully human antibodies. In one embodiment, the anti-PD-L1 antibody is an antibody produced by a mouse-derived hybridoma cell line. Thus, in one embodiment, the anti-PD-L1 antibody is a murine antibody. In another embodiment, the anti-PD-L1 antibody is a chimeric antibody. In another embodiment, the chimeric antibody is a mouse-human chimeric antibody. In another embodiment, the antibody is a humanized antibody. In another embodiment, the antibody is derived from a murine antibody and is humanized.
"humanized antibodies" are the following antibodies: the antibodies contain Complementarity Determining Regions (CDRs) derived from a non-human antibody; and framework and constant regions derived from human antibodies. For example, an anti-PD-L1 antibody provided herein can comprise CDRs derived from one or more murine antibodies as well as human framework and constant regions. Thus, in one embodiment, the humanized antibodies provided herein bind to the same epitope on PD-L1 as the murine antibody from which the CDRs of the antibodies are derived. Exemplary humanized antibodies are provided herein. Additional anti-PD-L1 antibodies or variants thereof comprising heavy and light chain CDRs provided herein can be generated using any human framework sequences and are also included in the disclosure. In one embodiment, framework sequences suitable for use in the present disclosure include those framework sequences that are similar in structure to the framework sequences provided herein. Additional modifications may be made in the framework regions to improve the properties of the antibodies provided herein. Such additional framework modifications may include chemical modifications; point mutations to reduce immunogenicity or to remove T cell epitopes; or reverting the mutation to a residue in the original germline sequence. In some embodiments, such modifications include those corresponding to the mutations exemplified herein, including back mutations to germline sequences. For example, in one embodiment, one or more amino acids in the human framework regions of VH and/or VL of a humanized antibody provided herein are back mutated to corresponding amino acids in a parent murine antibody. For example, for the VH and VL of humanized 5G11 and humanized 13C5, several sites of framework amino acids of the above template human antibodies were back mutated to the corresponding amino acid sequences in the mouse 5G11 and 13C5 antibodies. In one embodiment, the amino acids at positions 53 and/or 60 and/or 67 of the light chain variable region are back mutated to the corresponding amino acids found at said positions in the mouse 5G11 or 13C5 light chain variable region. In another embodiment, the amino acids at positions 24 and/or 28 and/or 30 and/or 49 and/or 73 and/or 83 and/or 94 of the heavy chain variable region are back mutated to the corresponding amino acids found at said positions in the mouse 5G11 or 13C5 heavy chain variable region. In one embodiment, the humanized 5G11 antibody comprises a light chain variable region in which the amino acid at position 60 is mutated from Ser (S) to Asp (D) and the amino acid at position 67 is mutated from Ser (S) to Tyr (Y); and a heavy chain variable region wherein the amino acid at position 24 is mutated from Phe (F) to Val (V), the amino acid at position 49 is mutated from Ala (a) to Gly (G), the amino acid at position 73 is mutated from Thr (T) to Asn (N), and the amino acid at position 83 is mutated from Thr (T) to Asn (N). In one embodiment, the humanized 13C5 antibody comprises a light chain variable region in which the amino acid at position 53 is mutated from Tyr (Y) to Lys (K); and a heavy chain variable region, wherein the amino acid at position 28 is mutated from Thr (T) to Ile (I), the amino acid at position 30 is mutated from Ser (S) to Arg (R), the amino acid at position 49 is mutated from Ser (S) to Ala (a), and the amino acid at position 94 is mutated from Tyr (Y) to Asp (D). Additional or alternative back mutations may be made in the framework regions of the humanized antibodies provided herein to improve the properties of the antibodies. The disclosure also includes humanized antibodies that bind PD-L1 and comprise framework modifications corresponding to the exemplary modifications described herein relative to any suitable framework sequence, as well as other framework modifications that otherwise improve the properties of the antibodies.
The present disclosure provides isolated antibodies or fragments thereof that bind PD-L1, wherein the antibodies can be produced by a hybridoma selected from the group consisting of hybridomas referred to herein as 13C5, 5G 11. Thus, the disclosure also includes hybridomas 13C5, 5G11, as well as any hybridomas that produce the antibodies disclosed herein. The disclosure also provides isolated polynucleotides encoding the antibodies and fragments thereof provided herein. The disclosure also includes expression vectors comprising the isolated polynucleotides, and host cells comprising the expression vectors.
"isolated antibody" means an antibody that: which is substantially free of other antibodies having different antigen specificities (e.g., an isolated antibody that specifically binds PD-1 is substantially free of antibodies that specifically bind antigens other than PD-1). However, an isolated antibody that specifically binds PD-1 may have cross-reactivity with other antigens (such as PD-1 molecules from different species). In addition, the isolated antibodies may be substantially free of other cellular material and/or chemicals.
The term "monoclonal antibody" ("mAb") refers to a non-naturally occurring preparation of antibody molecules of single molecular composition (i.e., antibody molecules whose basic sequences are substantially identical and which exhibit a single binding specificity and affinity for a particular epitope). A mAb is one example of an isolated antibody. Mabs may be produced by hybridoma techniques, recombinant techniques, transgenic techniques, or other techniques known to those skilled in the art.
The antibodies and antigen binding fragments thereof disclosed herein are specific for PD-L1. In one embodiment, the antibody or fragment thereof is specific for PD-L1. In one embodiment, the antibodies and fragments provided herein bind to human or primate PD-L1, but do not bind to PD-L1 from any other mammal. In another embodiment, the antibody or fragment thereof does not bind to mouse PD-L1. The terms "human PD-L1", "hPD-L1" and "huPD-L1" and the like are used interchangeably herein and refer to human PD-L1 and variants or isoforms of human PD-L1. By "specific" is meant that the antibody and fragments thereof bind PD-L1 with greater affinity than any other target.
The term "treatment" generally refers to obtaining a desired pharmacological and/or physiological effect. The effect may be therapeutic in terms of partial or complete stabilization or cure of the disease and/or side effects due to the disease. As used herein, "treatment" encompasses any treatment of a disease in a patient, including: (a) inhibiting the symptoms of the disease, i.e., arresting its development; or (b) alleviating a symptom of the disease, i.e., causing regression of the disease or symptom.
The term "preventing" means that a compound or formulation described in the present disclosure is administered to prevent a disease or one or more symptoms associated with the disease, including: preventing a disease or a disease state from occurring in a mammal, particularly when such mammal is susceptible to the disease state, but has not been diagnosed as having the disease state.
The term "effective amount" means an amount of a compound of the present disclosure that (i) treats a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The amount of active agent (e.g., fusion protein or compound of the present disclosure) that comprises a "therapeutically effective amount" can vary depending on factors such as the disease state, age, sex, and weight of the individual, and the ability of the therapeutic agent or combination of therapeutic agents to elicit a desired response in the individual. An effective amount can also be routinely determined by one of ordinary skill in the art based on its own knowledge and disclosure.
The term "administering" means physically introducing a composition comprising a therapeutic agent into a subject using any of a variety of methods and delivery systems known to those of skill in the art. Routes of administration of fusion proteins (e.g., anti-PD-L1 antibody-tgfbetarii fusion proteins) include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, such as by injection or infusion. The phrase "parenteral administration" as used herein refers to modes of administration other than enteral and topical administration, typically by injection, and includes, but is not limited to, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra-articular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, and in vivo electroporation. Administration may also be performed, for example, one, multiple times, and/or over one or more extended periods of time.
Unless otherwise indicated, the use of the term "dose" refers to the dose administered to a patient irrespective of the weight or Body Surface Area (BSA) of the patient. For example, 60kg of a human and 100kg of a human will receive the same dose of fusion protein.
The term "weight-based dose" as referred to herein refers to the dose calculated based on the weight of the patient that is administered to the patient. For example, when a patient having a body weight of 60kg requires 3mg/kg of fusion protein, one can withdraw an appropriate amount of fusion protein (i.e., 180 mg) at a time from a fixed dose pharmaceutical composition of fusion protein.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt" includes salts of a base ion with a free acid or salts of an acid ion with a free base.
The terms "subject" or "patient" are used interchangeably herein. In some embodiments, the term "subject" or "patient" is a mammal. In some embodiments, the subject or patient is a mouse. In some embodiments, the subject or patient is a human.
The term "about" is to be understood to include within three standard deviations of the average value or within standard tolerances in the particular field. In certain embodiments, a variation of no more than about 0.5 is understood. "about" modifies all values recited thereafter. For example, "about 1, 2, 3" means "about 1", "about 2", "about 3".
As used herein, "combined" or "combined" means that two or more active substances can each be administered to a subject simultaneously as a single formulation, or each as a single formulation sequentially in any order.
The term "single dose" refers to the smallest packaging unit containing a quantity of a drug, e.g., a box of seven capsules, each capsule being a single dose; or a single dose per bottle of injectate. The terms "single dose" and "unit dose" have the same meaning and are used interchangeably herein.
The term "multi-dose" consists of a plurality of single doses.
The term "daily dose" refers to the dose administered to a patient daily.
The term "daily dose" refers to the dose administered to a patient a day.
The term "pharmaceutical composition" refers to a mixture of one or more active ingredients of the present disclosure or pharmaceutical combinations thereof and pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate administration of a compound of the present disclosure, or a pharmaceutical combination thereof, to a subject.
The terms "day", "daily" and the like, when referring to a dosing regimen, refer to a time within a calendar day, beginning at midnight and ending at the next midnight.
In this document, the terms "comprises," "comprising," and "includes," or equivalents thereof, unless otherwise specified, are open ended and mean that other unspecified elements, components, and steps are contemplated in addition to those listed.
All patents, patent applications, and other identified publications are expressly incorporated herein by reference for the purpose of description and disclosure. These publications are provided solely for their disclosure prior to the filing date of the present disclosure. All statements as to the date or representation as to the contents of these documents are based on the information available to the applicant and do not constitute any admission as to the correctness of the dates or contents of these documents. Moreover, any reference to such publications in this document does not constitute an admission that the publications are part of the common general knowledge in the art, in any country.
Detailed Description
For clarity, the disclosure is further illustrated with examples, but the examples do not limit the scope of the disclosure. All reagents used in the present disclosure are commercially available and can be used without further purification.
Experimental example 1 in vitro cell proliferation inhibitory Activity
The antiproliferative potency of compounds of formula (I) and Abemaciclib on Rb gene normal cells was tested in parallel using celltiter glo technique to inhibit cell proliferation at a concentration of 0.3 μm for 72 hours.
The results of anti-cell proliferation are shown in Table 2.
TABLE 2
Experimental example 2 pharmacodynamics evaluation of Compound of formula (I) in MKN45 human gastric cancer nude mice subcutaneous transplantation tumor model
On the right armpit of SPF-class female nude mice (source: changzhou Kavens laboratory animal Co., ltd.) were inoculated with MKN45 cells subcutaneously, 5X 10 6 Individual/only (1:1 hybrid Matrigel). The average volume of the tumor is 100mm 3 When left and right, animals are grouped.
The day of grouping was day 0, and administration was performed once daily by intragastric administration starting on day 0. Tumor volume is measured for 2-3 times every week, meanwhile, the weight of the mice is weighed, and data are recorded; mice were observed and recorded daily for general performance. After the experiment is finished, the tumors are peeled, weighed and photographed.
The detection index and the calculation formula are as follows:
tumor volume, TV (mm) 3 )=1/2×(a×b 2 ) The method comprises the steps of carrying out a first treatment on the surface of the Wherein a is the long diameter of the tumor, and b is the short diameter of the tumor.
Rtv=tv relative to tumor volume t /TV 0 The method comprises the steps of carrying out a first treatment on the surface of the Wherein, TV 0 For tumor volume on day 0, TV t Tumor volume at each measurement.
Relative tumor proliferation rate, T/C (%) =t RTV /C RTV X 100%; wherein T is RTV RTV for treatment group; c (C) RTV RTV was vehicle control.
Tumor growth inhibition, TGI (%) = (1-TW/TW) 0 ) X 100%; wherein TW is the treatment of the tumor weight, TW 0 Tumor weight was determined as vehicle control.
Weight change rate, WCR (%) = (Wt) t -Wt 0 )/Wt 0 X 100%; wherein Wt is 0 Weight of the mice on day 0, wt t The body weight of the mice at each measurement was determined.
Experimental example 3 pharmacodynamic evaluation of Compounds of formula (I) in the model of subcutaneous transplantation tumor in SW620 human colon cancer nude mice
SW620 cells were inoculated subcutaneously in the right armpit of SPF-grade female nude mice (source: evergreen laboratory animal limited), 5×10 6 Individual/only (1:1 hybrid Matrigel). The average volume of the tumor is 100mm 3 When left and right, animals are grouped.
The day of grouping was day 0, and administration was performed once daily by intragastric administration starting on day 0. Tumor volume is measured for 2-3 times every week, meanwhile, the weight of the mice is weighed, and data are recorded; mice were observed and recorded daily for general performance. After the experiment is finished, the tumors are peeled, weighed and photographed.
The detection index and the calculation formula are as follows:
tumor volume, TV (mm) 3 )=1/2×(a×b 2 ) The method comprises the steps of carrying out a first treatment on the surface of the Wherein a is the long diameter of the tumor, and b is the short diameter of the tumor.
Rtv=tv relative to tumor volume t /TV 0 The method comprises the steps of carrying out a first treatment on the surface of the Wherein, TV 0 For tumor volume on day 0, TV t Tumor volume at each measurement.
Relative tumor proliferation rate, T/C (%) =t RTV /C RTV X 100%; wherein T is RTV RTV for treatment group; c (C) RTV RTV was vehicle control.
Tumor growth inhibition, TGI (%) = (1-TW/TW) 0 ) X 100%; wherein TW is the treatment of the tumor weight, TW 0 Tumor weight was determined as vehicle control.
Weight change rate, WCR (%) = (Wt) t -Wt 0 )/Wt 0 X 100%; wherein Wt is 0 Weight of the mice on day 0, wt t The body weight of the mice at each measurement was determined.
Experimental example 4 clinical trial
The study is the effectiveness and safety of the compound of formula (I) combined with an anti-PD-L1 antibody for treating PD-1/PD-L1 monoclonal antibody resistance and cell cycle abnormal digestive system tumor.
1.1 inclusion criteria:
1) Voluntary participation and written signing of informed consent;
2) Age is more than or equal to 18 years old;
3) The sex is unlimited;
4) Pathologically well-diagnosed digestive system tumor patients (including: esophageal cancer, gastric cancer, colorectal cancer, liver cancer);
5) Digestive system tumor patients who failed standard treatment;
6) Patients who failed treatment with PD-1 mab or PD-L1 mab;
7) There are cell cycle signal pathway abnormalities (e.g.: high expression of Cyclin D1, CDK4/6 mutation, etc.);
8) Imaging confirmed unresectable digestive system tumor patients;
9) At least one measurable focus (according to RECIST1.1 standard) or an assayable non-measurable focus, the imaging diagnosis is less than or equal to 21 days from the time of enrollment;
10 Predicted survival is greater than or equal to 3 months;
11 General physical condition (ECOG) 0-2;
12 Adequate bone marrow hematopoietic function (within 7 days): hemoglobin is more than or equal to 9g/dL, and leucocyte is more than or equal to 3.0X10 9 L/L the process comprises, neutrophils are more than or equal to 1.50X10 9 The ratio of the total amount of the components to the total amount of the components is greater than or equal to 100 multiplied by 10 9 L; liver and kidney function is normal (within 14 days): TBIL is less than or equal to 1.5 times of the upper limit of the normal value; ALT and AST are less than or equal to 2.5 times of the upper limit of the normal value, and if liver metastasis is caused, the ALT and AST are less than or equal to 5 times of the upper limit of the normal value; creatinine is less than or equal to 1.5 times of the upper limit of the normal value;
13 Heart, lung, kidney, liver function is approximately normal, laboratory tests meet the following criteria: urine routine inspection criteria: urine routine prompts urine protein < ++; if the urine protein is more than or equal to++, the urine protein ration is less than or equal to 1.0g after 24 hours. Blood clotting function check criteria: prothrombin Time (PT), activated Partial Thromboplastin Time (APTT), international Normalized Ratio (INR) less than or equal to 1.5 XULN (not receiving anticoagulation treatment);
14 Women of childbearing age should agree that effective contraceptive measures must be taken during the study period and within 6 months after the end of the study, and that serum or urine pregnancy tests are negative within 7 days prior to study entry into the group; men should agree that effective contraceptive measures must be taken during the study period and within 6 months after the end of the study period.
1.2 test drug
A compound capsule of formula (I): is provided by the pharmaceutical company of the pharmaceutical industry group, inc. Specification of: 5 mg/grain, 50 mg/grain.
anti-PD-L1 antibody injection hu5G11-hIgG1:1200mg: is provided by the pharmaceutical company of the pharmaceutical industry group, inc. Specification of: 100mg/10mL, 600mg/20mL.
1.3 dosing regimen
A compound capsule of formula (I): 120mg or 150mg or 180mg, is orally taken on an empty stomach, once daily, and continuously taken for 21 days as a treatment period. 120mg of subjects in group 1, if no DLT occurred, enter 150mg group, using 3+3 design; if DLT occurs, the 120mg group is converted to a 3+3 design. If DLT appears in patients with dose level group of 150mg and more than or equal to 2/6, 120mg is recommended as safe dose; if less than or equal to 1/6 of the patients show DLT, the DLT can be increased to 180mg. If the patient with the dose level of 180mg appears DLT, the patient with the dose level of less than or equal to 1/6 is considered to be a safe dose, and if the patient with the dose level of more than or equal to 2/6 appears DLT, 150mg is finally recommended to be the safe dose.
anti-PD-L1 antibody injection hu5G11-hIgG1:1200mg injection is diluted to 250mL by normal saline, and the infusion time is 60+/-10 min. Once every 21 days, the maximum use time is not more than 24 months.
1.4 evaluation criteria
Validity evaluation criteria: and evaluating the curative effect according to the RECIST 1.1 solid tumor alleviation evaluation standard.
Safety evaluation criteria: the severity of adverse events was judged using the NCI-CTC AE 5.0 standard.
1.5 results
Table 3 is a representative case, and the results show that the pharmaceutical combination of the compound capsule of formula (I) and the anti-PD-L1 antibody injection hu5G11-hIgG1 has significant clinical benefit in the aspect of treating digestive tract tumors.
TABLE 3 Table 3
/>

Claims (10)

1. A pharmaceutical combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and an anti-PD-L1 antibody
2. The pharmaceutical combination of claim 1, wherein the anti-PD-L1 antibody comprises the amino acid sequence: a heavy chain CDR1 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 1 or SEQ ID NO. 4; a heavy chain CDR2 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 2 or SEQ ID NO. 5; a heavy chain CDR3 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 3 or SEQ ID NO. 6; a light chain CDR1 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 7 or SEQ ID NO. 10; a light chain CDR2 region having at least 80% homology to the amino acid sequence shown in SEQ ID NO. 8 or SEQ ID NO. 11; a light chain CDR3 region having at least 80% homology with the amino acid sequence shown in SEQ ID NO. 9 or SEQ ID NO. 12.
3. The pharmaceutical combination of claim 1, comprising a pharmaceutical composition of a compound of formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutical composition of an anti-PD-L1 antibody.
4. A pharmaceutical combination according to any one of claims 1 to 3, which comprises 20 to 240mg, 40 to 180mg, 60 to 180mg, 80 to 180mg, 100 to 180mg, 120 to 180mg or 150 to 180mg of a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, calculated as a compound of formula (I); alternatively, it contains 20mg, 40mg, 60mg, 80mg, 100mg, 120mg, 150mg and/or 240mg of the compound of formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof, calculated as the compound of formula (I).
5. The pharmaceutical combination according to claim 4, wherein the pharmaceutical composition of the compound of formula (I) is a single dose or a plurality of doses, preferably the pharmaceutical composition of the compound of formula (I) is a plurality of doses.
6. The pharmaceutical combination according to any one of claims 1-3, comprising 600-2400 mg of an anti-PD-L1 antibody or a pharmaceutical composition thereof;
alternatively, it contains 600 to 2100mg, or 900 to 1500mg of the pharmaceutical composition of the anti-PD-L1 antibody.
7. The pharmaceutical combination of claim 6, wherein the pharmaceutical composition of the compound of formula (II) is a multi-dose.
8. A pharmaceutical combination according to any one of claims 1-3, which is a pharmaceutical composition suitable for administration in a single treatment cycle, comprising the following components in weight ratios (1.0-6.5), respectively: 1. preferably (2.0-6.5): 1, more preferably (2.0-3.5): 1, more preferably (2.1-3.2): 1, most preferably (2.6-3.2): 1, a compound of formula (I) or a pharmaceutically acceptable salt thereof, and an anti-PD-L1 antibody.
9. A pharmaceutical combination according to any one of claims 1 to 3, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is present in a daily dose; alternatively, the compound of formula (I) or a pharmaceutically acceptable salt thereof is present in an amount of once daily dose.
10. Use of a pharmaceutical combination according to any one of claims 1-8 for the manufacture of a medicament for the treatment or prevention of a tumor of the digestive system.
CN202311064786.9A 2022-08-23 2023-08-23 Combination pharmaceutical composition of CDK4/6 inhibitor and anti-PD-L1 antibody Pending CN117815387A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211014692 2022-08-23
CN2022110146926 2022-08-23

Publications (1)

Publication Number Publication Date
CN117815387A true CN117815387A (en) 2024-04-05

Family

ID=90517881

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311064786.9A Pending CN117815387A (en) 2022-08-23 2023-08-23 Combination pharmaceutical composition of CDK4/6 inhibitor and anti-PD-L1 antibody

Country Status (1)

Country Link
CN (1) CN117815387A (en)

Similar Documents

Publication Publication Date Title
CN113347996B (en) Combined pharmaceutical composition for treating tumors
CN113613674A (en) Combined pharmaceutical composition for treating small cell lung cancer
WO2020249018A1 (en) Combined pharmaceutical composition for treating driver-gene-positive lung cancer
CN113939315B (en) Combined pharmaceutical composition for treating melanoma
CN113811298B (en) Quinoline derivatives for the combined treatment of small cell lung cancer
CN113018429A (en) Pharmaceutical composition for treating ovarian cancer
CN112168961A (en) Combined pharmaceutical composition for treating colorectal cancer
CN115151258A (en) Combination cancer therapy using PD-1 antagonists, ILT4 antagonists and lenvatinib or a salt
WO2021219138A1 (en) Combination drug for treating kidney cancer
CN117815387A (en) Combination pharmaceutical composition of CDK4/6 inhibitor and anti-PD-L1 antibody
WO2023198060A1 (en) Pharmaceutical combination of proteasome inhibitor and anti-pd-1 antibody
WO2023174408A1 (en) Pharmaceutical combination of anti-tim-3 antibody and anti-pd-l1 antibody
US20230062308A1 (en) Treatment of ck8 positive cancers in relation with k-ras gene status
WO2023174278A1 (en) Pharmaceutical composition of anti-tim-3 antibody and hypomethylating agent
WO2023134787A2 (en) Use of combination of anti-pd-1 antibody and anti-vegf antibody in treatment of hepatocellular carcinoma
WO2024041652A1 (en) Methods of cancer treatment
CN116209443A (en) Pharmaceutical composition for treating small cell lung cancer
CN117642181A (en) Pharmaceutical composition for treating esophageal cancer
CN116370641A (en) Combined medicine for treating digestive system malignant tumor
TW202346342A (en) Drug combination of anti-TIM-3 antibody and anti-PD-1 antibody
CN115957321A (en) Application of anti-HER 2 antibody in preparation of medicine for treating cancer
CN116036265A (en) Combination medicine for cancers
CN117085124A (en) Pharmaceutical composition comprising anti-PD-L1 antibody and c-Met kinase inhibitor
CN116437957A (en) Combination pharmaceutical composition of anti-PD-L1 antibody and c-Met kinase inhibitor for treating lung cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication